Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Subcell Biochem ; 45: 365-83, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18193644

RESUMO

The plasma membrane calcium ATPase (PMCA) uses energy to pump calcium (Ca2+) ions out of the cytosol into the extracellular milieu, usually against a strong chemical gradient. This energy expenditure is necessary to maintain a relatively low intracellular net Ca2+ load. Mammals have four genes (ATP2B1-ATP2B4), encoding the proteins PMCA1 through PMCA4. Transcripts from each of these genes are alternatively spliced to generate several variant proteins that are in turn post-translationally modified in a variety of ways. Expressed ubiquitously and with some level of functional redundancy in most vital tissues, only one of the four genes--Atp2b2--has been causally linked through naturally occuring mutations to disease in mammals: specifically to deafness and ataxia in spontaneous mouse mutants. In humans, a missense amino acid substitution in PMCA2 modifies the severity of hearing loss. Targeted null mutations of the Atp2b1 and Atp2b4 genes in mouse are embryonic lethal and cause a sperm motility defect, respectively. These phenotypes point to complex human diseases like hearing loss, cardiac function and infertility. Changes in PMCA expression are associated with other diseases including cataract formation, carciniogenesis, diabetes, and cardiac hypertension and hypertrophy. Severity of these diseases may be affected by subtle changes in expression of the PMCA isoforms expressed in those tissues.


Assuntos
Cardiomiopatias/fisiopatologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/fisiologia , Animais , Humanos , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética
2.
Neuroscience ; 141(1): 245-57, 2006 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16675132

RESUMO

Atp2b2 encodes the plasma membrane Ca(2+)-ATPase type 2 (PMCA2) expressed in various tissues, including stereocilia of cochlear and vestibular hair cells, cerebellar Purkinje cells, and lactating mammary epithelia. Mutations of the gene lead to deafness, ataxia, and reduced Ca(2+) levels in milk. Heterozygous mutants also have abnormal hearing, suggesting that precise regulation of Atp2b2 is required for normal function. In this study, we describe Atp2b2 5'-untranslated region genomic structure and transcript usage in mice. Using 5'-rapid amplification of cDNA ends, we observed four transcripts: types alpha, beta, mu and delta, each splicing into a common ATG-containing exon. Types alpha and beta correspond to previously published mammalian cDNA sequences. Types mu and delta constitute novel 5'-untranslated region sequences, and were observed at high levels only in lactating mammary gland. Using real-time reverse transcriptase polymerase chain reaction, we quantified relative transcript usage across several tissues. We show that alpha and beta are abundant throughout the CNS, as well as the cochlea. When we microdissected the cochlea into hair cell and spiral ganglion containing fractions, we found that cochlear hair cell expression is mediated through the type alpha transcript. In situ hybridization studies in cerebellum using exon-specific probes revealed that alpha dominates in Purkinje neurons, while beta is enriched in cerebellar granule neurons. We compared 5'-untranslated region sequence across multiple species, and found high conservation around the first exons for alpha and beta in mammals, but not other species. The regions around the mu and delta first exons are highly conserved between rat and mouse, but less so with other species. Our results show that expression of Atp2b2 is highly regulated, using four different transcriptional start regions, two of which are differentially expressed in neuronal tissue. This suggests that unique regulatory mechanisms are used to control Atp2b2 expression in different types of cells.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Éxons/fisiologia , Células Ciliadas Auditivas/metabolismo , Glândulas Mamárias Animais/metabolismo , Neurônios/metabolismo , Regiões 5' não Traduzidas/fisiologia , Animais , Animais Recém-Nascidos , ATPases Transportadoras de Cálcio/genética , Proteínas de Transporte de Cátions/genética , Feminino , Expressão Gênica/fisiologia , Células Ciliadas Auditivas/crescimento & desenvolvimento , Hibridização In Situ/métodos , Masculino , Glândulas Mamárias Animais/crescimento & desenvolvimento , Camundongos , Camundongos Endogâmicos CBA , ATPases Transportadoras de Cálcio da Membrana Plasmática , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transcrição Gênica
3.
Epilepsy Res ; 66(1-3): 75-90, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16144756

RESUMO

PURPOSE: To develop a non-invasive method for exploring seizure initiation and propagation in the brain of intact experimental animals. METHODS: We have developed and applied a model-independent statistical method--Hierarchical Cluster Analysis (HCA)--for analyzing BOLD-fMRI data following administration of pentylenetetrazol (PTZ) to intact rats. HCA clusters voxels into groups that share similar time courses and magnitudes of signal change, without any assumptions about when and/or where the seizure begins. RESULTS: Epileptiform spiking activity was monitored by EEG (outside the magnet) following intravenous PTZ (IV-PTZ; n=4) or intraperitoneal PTZ administration (IP-PTZ; n=5). Onset of cortical spiking first occurred at 29+/-16 s (IV-PTZ) and 147+/-29 s (IP-PTZ) following drug delivery. HCA of fMRI data following IV-PTZ (n=4) demonstrated a single dominant cluster, involving the majority of the brain and first activating at 27+/-23s. In contrast, IP-PTZ produced multiple, relatively small, clusters with heterogeneous time courses that varied markedly across animals (n=5); activation of the first cluster (involving cortex) occurred at 130+/-59 s. With both routes of PTZ administration, the timing of the fMRI signal increase correlated with onset of EEG spiking. CONCLUSIONS: These experiments demonstrate that fMRI activity associated with seizure activity can be analyzed with a model-independent statistical method. HCA indicated that seizure initiation in the IV- and IP-PTZ models involves multiple regions of sensitivity that vary with route of drug administration and that show significant variability across animal subjects. Even given this heterogeneity, fMRI shows clear differences that are not apparent with typical EEG monitoring procedures, in the activation patterns between IV and IP-PTZ models. These results suggest that fMRI can be used to assess different models and patterns of seizure activation.


Assuntos
Encéfalo , Imageamento por Ressonância Magnética , Pentilenotetrazol , Convulsões/fisiopatologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Mapeamento Encefálico , Análise por Conglomerados , Modelos Animais de Doenças , Eletroencefalografia/efeitos dos fármacos , Processamento de Imagem Assistida por Computador/métodos , Masculino , Oxigênio/sangue , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente
4.
Neuroscience ; 119(1): 65-72, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12763069

RESUMO

Prostaglandin E(2) (PGE(2)) enhances the sensitivity of sensory neurons to various forms of noxious stimulation. This occurs, in part, by the suppression of a delayed rectifier-like potassium current in these neurons. However, the molecular identity of this current remains unclear. Recent studies demonstrated that a mutant mouse lacking a delayed rectifier potassium channel gene, Kv1.1, displayed lowered thresholds to thermal stimulation in behavioral assays of pain perception, i.e. the Kcna1-null mice were hyperalgesic. Here we examined whether PGE(2) can alter the sensitivity of Kcna1-null mice to noxious stimulation and examine the capability of PGE(2) to inhibit the potassium current in these knockout mice. Behavioral assays were used to assess the effect of PGE(2) on either thermal hyperalgesia or mechanical sensitivities. In addition, the whole-cell patch-clamp technique was used to study the effects of PGE(2) on the total potassium current recorded from isolated mouse sensory neurons. Even with a reduced threshold to thermal stimulation, PGE(2) could still sensitize the response of Kcna1-null mice to thermal and mechanical stimulation by amounts that were similar to that in wild type mice. The activation properties of the potassium current were similar for both the wild type and the Kcna1-null mice, whereas the inactivation properties were different in cells exhibiting large amounts of steady-state inactivation (>50%) measured at +20 mV. PGE(2) suppressed the total potassium current in both groups of mice by 40-50% without altering the voltage dependence of activation. In addition, PGE(2) produced similar amounts of suppression in both groups of mice when currents were examined with the steady-state inactivation protocol. Based on these results, it is unlikely that Kv1.1 is the molecular identity of the potassium channel(s) modulated by PGE(2) to sensitize nociceptive sensory neurons. Also, the enhanced thermal sensitivity as observed in the Kcna1-null mice might be due to more central neurons of the pain sensing pathway.


Assuntos
Dinoprostona/farmacologia , Neurônios Aferentes/efeitos dos fármacos , Ocitócicos/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/metabolismo , Animais , Comportamento Animal , Células Cultivadas , Condutividade Elétrica , Técnicas Eletrofisiológicas Cardíacas , Genótipo , Hiperalgesia , Canal de Potássio Kv1.1 , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout/genética , Camundongos Knockout/fisiologia , Estimulação Física , Canais de Potássio/deficiência , Canais de Potássio/genética , Tempo de Reação , Limiar Sensorial/efeitos dos fármacos , Fatores de Tempo
5.
Neurology ; 60(1): 22-6, 2003 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-12525712

RESUMO

BACKGROUND: Charcot-Marie-Tooth (CMT) neuropathy is a heterogeneous group of inherited disorders of the peripheral nervous system. The authors recently mapped an autosomal dominant demyelinating form of CMT type 1 (CMT1C) to chromosome 16p13.1-p12.3. OBJECTIVE: To find the gene mutations underlying CMT1C. METHODS: The authors used a combination of standard positional cloning and candidate gene approaches to identify the causal gene for CMT1C. Western blot analysis was used to determine relative protein levels in patient and control lymphocyte extracts. Northern blotting was used to characterize gene expression in 1) multiple tissues; 2) developing sciatic nerve; and 3) nerve-crush and nerve-transection experiments. RESULTS: The authors identified missense mutations (G112S, T115N, W116G) in the LITAFgene (lipopolysaccharide-induced tumor necrosis factor-alpha factor) in three CMT1C pedigrees. LITAF, which is also referred to as SIMPLE, is a widely expressed gene encoding a 161-amino acid protein that may play a role in protein degradation pathways. The mutations associated with CMT1C were found to cluster, defining a domain of the LITAF protein having a critical role in peripheral nerve function. Western blot analysis suggested that the T115N and W116G mutations do not alter the level of LITAF protein in peripheral blood lymphocytes. The LITAF transcript is expressed in sciatic nerve, but its level of expression is not altered during development or in response to nerve injury. This finding is in stark contrast to that seen for other known genes that cause CMT1. CONCLUSIONS: Mutations in LITAF may account for a significant proportion of CMT1 patients with previously unknown molecular diagnosis and may define a new mechanism of peripheral nerve perturbation leading to demyelinating neuropathy.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas de Membrana , Mutação de Sentido Incorreto , Proteínas Nucleares , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Cromossomos Humanos Par 16/genética , Clonagem Molecular , Análise Mutacional de DNA , Feminino , Regulação da Expressão Gênica , Testes Genéticos , Humanos , Masculino , Dados de Sequência Molecular , Regeneração Nervosa/genética , Especificidade de Órgãos , Linhagem , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína/genética , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/crescimento & desenvolvimento , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Fatores de Transcrição/biossíntese
6.
Nat Genet ; 29(1): 61-5, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11528393

RESUMO

The autosomal recessive mouse mutation quivering (qv), which arose spontaneously in 1953, produces progressive ataxia with hind limb paralysis, deafness and tremor. Six additional spontaneous alleles, qvJ, qv2J, qv3J, qv4J, qvlnd and qvlnd2J, have been identified. Ear twitch responses (Preyer's reflex) to sound are absent in homozygous qv/qv mice, although cochlear morphology seems normal and cochlear potentials recorded at the round window are no different from those of control mice. However, responses from brainstem auditory nuclei show abnormal transmission of auditory information, indicating that, in contrast to the many known mutations causing deafness originating in the cochlea, deafness in qv is central in origin. Here we report that quivering mice carry loss-of-function mutations in the mouse beta-spectrin 4 gene (Spnb4) that cause alterations in ion channel localization in myelinated nerves; this provides a rationale for the auditory and motor neuropathies of these mice.


Assuntos
Surdez/genética , Mutação , Espectrina/genética , Tremor/genética , Animais , Córtex Auditivo/fisiopatologia , Genes Recessivos , Camundongos , Camundongos Endogâmicos CBA , Camundongos Mutantes , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Genomics ; 74(3): 306-12, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11414758

RESUMO

The electroconvulsive threshold (ECT) test is used commonly in the screening of anti-epileptic drugs in rodent models, but little is known about its genetic or mechanistic basis. Thresholds for minimal clonic, maximal tonic, or psychomotor (partial) seizures were determined in 16 different inbred mouse strains in two different laboratories. A wide range of thresholds was observed, suggesting that a variety of neuroexcitability alleles exist in inbred strains. Although there was generally good cross-strain correlation between the three seizure types, several outlier strains were detected, showing that genetically encoded differences can affect the ability of a particular seizure type to spread through the brain. Furthermore, the relative seizure susceptibility of a strain was comparable between the two laboratories, suggesting that despite different test sites, instrumentation, and personnel, the ECT assay is portable and that common inbred strains can often be relied upon as calibration standards. Last, the ECT paradigm was also sensitive enough to detect single locus differences, laying the groundwork for mutation screens for new neuroexcitability models.


Assuntos
Canais de Potássio de Abertura Dependente da Tensão da Membrana , Convulsões/fisiopatologia , Animais , Eletrochoque , Genótipo , Canal de Potássio Kv1.1 , Masculino , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Canais de Potássio/genética , Convulsões/genética , Especificidade da Espécie
8.
J Physiol ; 533(Pt 3): 697-710, 2001 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-11410627

RESUMO

1. Ventricular myocytes demonstrate a steeply inwardly rectifying K(+) current termed I(K1). We investigated the molecular basis for murine I(K1) by removing the genes encoding Kir2.1 and Kir2.2. The physiological consequences of the loss of these genes were studied in newborn animals because mice lacking Kir2.1 have a cleft palate and die shortly after birth. 2. Kir2.1 (-/-) ventricular myocytes lack detectable I(K1) in whole-cell recordings in 4 mM external K(+). In 60 mM external K(+) a small, slower, residual current is observed. Thus Kir2.1 is the major determinant of I(K1). Sustained outward K(+) currents and Ba(2+) currents through L- and T-type channels were not significantly altered by the mutation. A 50 % reduction in I(K1) was observed in Kir2.2 (-/-) mice, raising the possibility that Kir2.2 can also contribute to the native I(K1). 3. Kir2.1 (-/-) myocytes showed significantly broader action potentials and more frequent spontaneous action potentials than wild-type myocytes. 4. In electrocardiograms of Kir2.1 (-/-) neonates, neither ectopic beats nor re-entry arrhythmias were observed. Thus the increased automaticity and prolonged action potential of the mutant ventricular myocytes were not sufficiently severe to disrupt the sinus pacing of the heart. The Kir2.1 (-/-) mice, however, had consistently slower heart rates and this phenotype is likely to arise indirectly from the influence of Kir2.1 outside the heart. 5. Thus Kir2.1 is the major component of murine I(K1) and the Kir2.1 (-/-) mouse provides a model in which the functional consequences of removing I(K1) can be studied at both cellular and organismal levels.


Assuntos
Miocárdio/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização , Canais de Potássio/fisiologia , Potenciais de Ação/fisiologia , Animais , Animais Recém-Nascidos , Cálcio/fisiologia , Condutividade Elétrica , Eletrocardiografia , Deleção de Genes , Ventrículos do Coração , Camundongos , Camundongos Endogâmicos , Camundongos Knockout/genética , Fenótipo , Canais de Potássio/genética , RNA/metabolismo , Tempo de Reação/fisiologia , Valores de Referência
9.
Neuroscience ; 103(4): 921-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11301201

RESUMO

Mice lacking the potassium channel subunit KCNA1 exhibit a severe epileptic phenotype beginning at an early postnatal age. The precise cellular physiological substrates for these seizures are unclear, as is the site of origin. Since KCNA1 mRNA in normal mice is expressed in the neocortex, we asked whether neurons in the neocortex of three to four week-old Kcna1-null mutants exhibit evidence of hyperexcitability. Layer V pyramidal neurons were directly visualized in brain slices with infrared differential-interference contrast microscopy and evaluated with cellular electrophysiological techniques. There were no significant differences in intrinsic membrane properties and action potential shape between Kcna1-null and wild-type mice, consistent with previous findings in hippocampal slice recordings. However, the frequency of spontaneous post-synaptic currents was significantly higher in Kcna1-null compared to wild-type mice. The frequency of spontaneous inhibitory post-synaptic currents and miniature (action-potential-independent) inhibitory post-synaptic currents was also significantly higher in Kcna1-null compared to wild-type mice. However, the frequency of spontaneous and miniature excitatory post-synaptic currents was not different in these two groups of animals. Comparison of the amplitude and kinetics of miniature inhibitory and excitatory post-synaptic currents revealed differences in amplitude, rise time and half-width between Kcna1-null and wild-type mice. Our data indicate that the inhibitory drive onto layer V pyramidal neurons is increased in Kcna1 knockout mice, either directly through an increased spontaneous release of GABA from presynaptic terminals contacting layer V pyramidal neurons, or an enhanced excitatory synaptic input to inhibitory interneurons.


Assuntos
Camundongos Knockout/genética , Camundongos Knockout/fisiologia , Inibição Neural/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Células Piramidais/fisiologia , Animais , Comportamento Animal/fisiologia , Condutividade Elétrica , Eletroencefalografia , Eletrofisiologia , Técnicas In Vitro , Canal de Potássio Kv1.1 , Camundongos , Camundongos Endogâmicos C3H , Convulsões/genética , Convulsões/fisiopatologia , Sinapses/fisiologia , Gravação de Videoteipe
10.
Hear Res ; 151(1-2): 205-220, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11124466

RESUMO

The deafwaddler (dfw) mouse mutant is caused by a spontaneous mutation in the gene that encodes a plasma membrane Ca(2+) ATPase (type 2), PMCA2 (Street et al., 1998. Nat. Genet. 19, 390-394), which is expressed in cochlear and vestibular hair cells. Distortion product otoacoustic emission (DPOAE) amplitudes and latencies were examined in control mice, deafwaddler mutants, and controls treated with the drug furosemide. Furosemide causes a transient reduction of DPOAEs (Mills et al., 1993. J. Acoust. Soc. Am. 94, 2108-2122). We wanted to determine whether DPOAEs obtained in furosemide-treated mice were similar or different from results obtained in +/dfw mice. DPOAE amplitude and phase were measured as a function of f(2)/f(1) ratio. These data were converted into waveforms using inverse fast Fourier transform, and their average latency was used to estimate DPOAE group delay. Homozygous deafwaddlers did not produce DPOAEs. Heterozygous deafwaddlers (+/dfw) had increased DPOAE thresholds and reduced amplitudes at high frequencies, compared to controls. To the extent that DPOAEs depend on functional outer hair cells (OHCs), abnormal DPOAEs in +/dfw mice suggest that PMCA2 is important for OHC function at high frequencies. Similar to the effects of furosemide, the mutation reduced DPOAEs for low-level stimuli; in contrast to furosemide, the mutation altered DPOAEs elicited by high levels.


Assuntos
ATPases Transportadoras de Cálcio/genética , Surdez/genética , Surdez/fisiopatologia , Mutação , Emissões Otoacústicas Espontâneas/genética , Emissões Otoacústicas Espontâneas/fisiologia , Animais , Proteínas de Transporte de Cátions , Surdez/enzimologia , Furosemida/toxicidade , Células Ciliadas Auditivas Externas/efeitos dos fármacos , Células Ciliadas Auditivas Externas/fisiopatologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Emissões Otoacústicas Espontâneas/efeitos dos fármacos , ATPases Transportadoras de Cálcio da Membrana Plasmática
11.
Ment Retard Dev Disabil Res Rev ; 6(4): 281-92, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11107193

RESUMO

Our understanding of the genetic basis of epilepsy is progressing at a rapid pace. Gene mutations causing several of the inherited epilepsies have been mapped, and several more are likely to be added in coming years. In this review, we summarize the available information on the genetic basis of human epilepsies and epilepsy syndromes, emphasizing how genetic defects may correlate with the pathophysiological mechanisms of brain hyperexcitability. Mutations leading to epilepsy have been identified in genes encoding voltage- and ligand-gated ion channels (benign familial neonatal convulsions, autosomal dominant nocturnal frontal lobe epilepsy, generalized epilepsy with febrile seizures "plus"), neurotransmitter receptors (Angelman syndrome), the molecular cascade of cellular energy production (myoclonic epilepsy with ragged red fibers), and proteins without a known role in neuronal excitability (Unverricht-Lundborg disease). Gene defects can lead to epilepsy by altering multiple and diverse aspects of neuronal function.


Assuntos
Epilepsia/genética , Animais , Causalidade , Mapeamento Cromossômico/estatística & dados numéricos , Metabolismo Energético/genética , Epilepsia/patologia , Epilepsia/fisiopatologia , Humanos , Canais Iônicos/genética , Mutação/fisiologia , Epilepsias Mioclônicas Progressivas/genética , Epilepsias Mioclônicas Progressivas/patologia , Epilepsias Mioclônicas Progressivas/fisiopatologia , Neurônios/metabolismo , Neurônios/patologia , Receptores de Neurotransmissores/genética , Receptores de Neurotransmissores/metabolismo
12.
Hear Res ; 140(1-2): 77-90, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10675636

RESUMO

Voltage-gated potassium (Kv) channels may play an important role in the encoding of auditory information. Towards understanding the roles of Shaker and Shaw-like channels in this process, we examine here the expression of Kv1.1, Kv1.2, Kv3.1, and Kv3.3 in the central auditory nuclei of the mouse using quantitative in situ hybridization techniques. We establish rank order for each channel's expression in each region, finding that the medial nucleus of the trapezoid body shows the highest signal for each of the four channel genes. In other auditory nuclei differential expression is found among and between members of both Shaker and Shaw subfamilies. Of particular interest is the stark contrast between high level expression of Kv1.1 and very low level expression of Kv3.1 in the octopus cell area of the cochlear nucleus and in the lateral superior olivary nucleus. These unique expression patterns suggest that Kv channel gene expression is regulated to allow brainstem auditory neurons to transmit temporally patterned signals with high fidelity. In instances where specific cell types can be tentatively identified, we discuss the possible contribution made by these channel genes to the physiological properties of those neurons.


Assuntos
Tronco Encefálico/metabolismo , Núcleo Coclear/metabolismo , Canais de Potássio/genética , Animais , Regulação da Expressão Gênica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
13.
Dev Neurosci ; 21(3-5): 320-7, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10575255

RESUMO

Potassium channels play a critical role in limiting neuronal excitability. Mutations in certain voltage-gated potassium channels have been associated with hyperexcitable phenotypes in both humans and animals. However, only recently have mutations in potassium channel genes (i.e. KCNQ2 and KCNQ3) been discovered in a human epilepsy, benign familial neonatal convulsions. Recently, it has been reported that mice lacking the voltage-gated Shaker-like potassium channel Kv1.1 alpha-subunit develop recurrent spontaneous seizures early in postnatal development. The clinical relevance of the Kv1.1 knockout mouse has been underscored by a recent report of epilepsy occurring in a family affected by mutations in the KCNA1 locus (the human homologue of Kv1.1) which typically cause episodic ataxia and myokymia. Here we summarize preliminary studies characterizing the developmental changes in seizure susceptibility and neuronal activation in the three genotypes of Kv1.1 mice (-/-, +/-, +/+). Using behavioral and immediate-early gene indicators of regional brain excitability, we have found that a seizure-sensitive predisposition exists in Kv1.1 -/- animals at a very young age (P10), before either spontaneous seizure activity or changes in c-fos mRNA expression can be demonstrated. Kv1.1 +/- mice, although behaviorally indistinguishable from wild types, also have an increased susceptibility to seizures at a similar early age. The Kv1. 1 knockout mouse possesses many features desirable in a developmental animal epilepsy model and represents a clinically relevant model of early-onset epilepsies.


Assuntos
Animais Recém-Nascidos/crescimento & desenvolvimento , Camundongos Knockout/genética , Camundongos Knockout/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Convulsões/etiologia , Animais , Suscetibilidade a Doenças , Canal de Potássio Kv1.1 , Camundongos , Camundongos Knockout/crescimento & desenvolvimento
15.
J Biol Chem ; 273(35): 22807-18, 1998 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-9712915

RESUMO

Potassium channels play important roles in shaping the electrical properties of excitable cells. Toward understanding the transcriptional regulation of a member of the inwardly rectifying potassium channel family, we have characterized the genomic structure and 5'-proximal promoter of the murine Kcnj2 gene (also referred to as IRK1 and Kir2.1). The Kcnj2 transcription unit is composed of two exons separated by a 5.5-kilobase pair intron. Deletion analysis of 5'-flanking sequences identified a promiscuously active 172-base pair minimal promoter, whereas expression from a construct containing additional upstream sequences was cell type-restricted. The minimal promoter contained an E box, a Y box, and three GC box consensus elements but lacked both TATA and CCAAT box elements. The activity of the minimal promoter was found to be controlled by a combination of the activities of the transcription factors Sp1, Sp3, and NF-Y. The interplay between Sp1, Sp3, and NF-Y within the architecture of the Kcnj2 promoter, the ubiquitous nature of these trans-acting factors, and the action of tissue-selective repressor element(s) may combine to enable a wide variety of cell types to differentially regulate Kcnj2 expression through transcriptional control.


Assuntos
Regulação da Expressão Gênica/genética , Canais de Potássio/genética , Regiões Promotoras Genéticas , Transcrição Gênica/genética , Animais , Sequência de Bases , Ligação Competitiva , Proteínas Estimuladoras de Ligação a CCAAT , Linhagem Celular , DNA Complementar , Proteínas de Ligação a DNA/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação Puntual , Canais de Potássio/metabolismo , Ligação Proteica , Fator de Transcrição Sp1/metabolismo , Fator de Transcrição Sp3 , Fatores de Transcrição/metabolismo
16.
Neurosci Lett ; 251(2): 121-4, 1998 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-9718989

RESUMO

Hyperalgesia and morphine induced antinociception were measured in mice lacking the gene for the Shaker-like voltage-gated potassium channel Kv1.1 alpha subunit. The effects of varying gene dosage were studied by comparing homozygous null (-/-) versus heterozygous (+/-) and wildtype (+/+) littermates. Hyperalgesia was measured using the paw flick assay, hot plate assay and formalin induced hind paw licking. It was observed that null mutant animals had significantly shorter latencies to response in the paw flick (36%) and hot plate (27%) assays while their licking times after hind paw injection of formalin was increased in both the first (74%) and second (65%) phases of the response compared to wildtype controls. Morphine induced antinociception in Kv1.1 null mutant animals was blunted. These studies indicate that Kv1.1 plays an important role in nociceptive and antinociceptive signaling pathways.


Assuntos
Hiperalgesia/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/deficiência , Canais de Potássio/genética , Animais , Hiperalgesia/metabolismo , Canal de Potássio Kv1.1 , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout
17.
Nat Genet ; 19(4): 390-4, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9697703

RESUMO

Hearing loss is the most common sensory deficit in humans. Because the auditory systems of mice and humans are conserved, studies on mouse models have predicted several human deafness genes and identified new genes involved in hearing. The deafwaddler (dfw) mouse mutant is deaf and displays vestibular/motor imbalance. Here we report that the gene encoding a plasma membrane Ca2+-ATPase type 2 pump (Atp2b2, also known as Pmca2) is mutated in dfw. An A-->G nucleotide transition in dfw DNA causes a glycine-to-serine substitution at a highly conserved amino-acid position, whereas in a second allele, dfw2J, a 2-base-pair deletion causes a frameshift that predicts a truncated protein. In the cochlea, the protein Atp2b2 is localized to stereocilia and the basolateral wall of hair cells in wild-type mice, but is not detected in dfw2J mice. This indicates that mutation of Atp2b2 may cause deafness and imbalance by affecting sensory transduction in stereocilia as well as neurotransmitter release from the basolateral membrane. These mutations affecting Atp2b2 in dfw and dfw2J are the first to be found in a mammalian plasma membrane calcium pump and define a new class of deafness genes that directly affect hair-cell physiology.


Assuntos
ATPases Transportadoras de Cálcio/genética , Surdez/genética , Mutação da Fase de Leitura/genética , Mutação Puntual/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Encéfalo/enzimologia , ATPases Transportadoras de Cálcio/análise , Membrana Celular/enzimologia , Mapeamento Cromossômico , Análise Mutacional de DNA , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Genes/genética , Camundongos , Camundongos Mutantes Neurológicos , Dados de Sequência Molecular , Órgão Espiral/enzimologia , RNA Mensageiro/análise , Vestíbulo do Labirinto/enzimologia
18.
J Neurosci ; 18(15): 5682-91, 1998 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-9671659

RESUMO

Kv1.1, a Shaker-like voltage-gated potassium channel, is strongly expressed in a variety of neurons in adult rodents, in which it appears to be involved in regulating neuronal excitability. Here we show that Kv1.1 is also expressed during embryonic development in the mouse, exhibiting two transient peaks of expression around embryonic day 9.5 (E9.5) and E14.5. Using both in situ hybridization and immunocytochemistry, we have identified several cell types and tissues that express Kv1.1 RNA and protein. At E9.5, Kv1.1 RNA and protein are detected transiently in non-neuronal cells in several regions of the early CNS, including rhombomeres 3 and 5 and ventricular zones in the mesencephalon and diencephalon. At E14.5, several cell types in both the CNS and peripheral nervous system express Kv1.1, including neuronal cells (sensory ganglia and outer aspect of cerebral hemispheres) and glial cells (radial glia, satellite cells, and Schwann cell precursors). These data show that Kv1.1 is expressed transiently in a variety of neuronal and non-neuronal cells during restricted periods of embryonic development. Although the functional roles of Kv1.1 in development are not understood, the cell-specific localization and timing of expression suggest this channel may play a role in several developmental processes, including proliferation, migration, or cell-cell adhesion.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , Animais , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário e Fetal/fisiologia , Idade Gestacional , Ativação do Canal Iônico , Canal de Potássio Kv1.1 , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Especificidade de Órgãos , RNA Mensageiro/metabolismo , Fatores de Tempo
19.
Proc Natl Acad Sci U S A ; 95(13): 7693-8, 1998 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-9636212

RESUMO

The tissue distributions and physiological properties of a variety of cloned voltage-gated potassium channel genes have been characterized extensively, yet relatively little is known about the mechanisms controlling expression of these genes. Here, we report studies on the regulation of Kv1.1 expressed endogenously in the C6 glioma cell line. We demonstrate that elevation of intracellular cAMP leads to the accelerated degradation of Kv1.1 RNA. The cAMP-induced decrease in Kv1.1 RNA is followed by a decrease in Kv1. 1 protein and a decrease in the whole cell sustained K+ current amplitude. Dendrotoxin-I, a relatively specific blocker of Kv1.1, blocks 96% of the sustained K+ current in glioma cells, causing a shift in the resting membrane potential from -40 mV to -7 mV. These data suggest that expression of Kv1.1 contributes to setting the resting membrane potential in undifferentiated glioma cells. We therefore suggest that receptor-mediated elevation of cAMP reduces outward K+ current density by acting at the translational level to destabilize Kv1.1 RNA, an additional mechanism for regulating potassium channel gene expression.


Assuntos
AMP Cíclico/fisiologia , Glioma/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , RNA Mensageiro/metabolismo , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Células CHO , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Venenos Elapídicos/farmacologia , Eletrofisiologia , Glioma/metabolismo , Humanos , Canal de Potássio Kv1.1 , Inibidores de Fosfodiesterase/farmacologia , Canais de Potássio/metabolismo , Células Tumorais Cultivadas
20.
Genomics ; 49(3): 371-7, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9615221

RESUMO

The deafwaddler (dfw) mutation, displaying motor ataxia and profound deafness, arose spontaneously in a C3H/HeJ colony and was mapped previously to distal mouse Chr 6. In this study, a high-resolution genetic map was generated by positioning 10 microsatellite markers and 5 known genes on a 968-meioses intersubspecific backcross segregating for dfw [(CAST/Ei(-)+/+ x C3HeB/ FeJ-dfw/dfw) x C3HeB/FeJ-dfw/dfw], giving the following marker order and sex-averaged distances: D6Mit64-(0.10 + 0.10 cM)-Pang-(1.24 + 0.36 cM)-Itpr1-(0.62 + 0.25 cM)-D6Mit108-(0.52 + 0.23 cM)-D6Mit54-(0.21 + 0.15 cM)-D6Mit23, D6Mit107, D6Mit328-(0.72 + 0.27 cM)-D6Mit11-(0.21 + 0.15 cM)-dfw-(0.93 + 0.31 cM)-Gat4, D6Mit55-(0.10 + 0.10 cM)-D6Mit63-(0.31 + 0.18 cM)-Syn2-(0.62 + 0.25 cM)-D6Mit44 (Rho). Female and male genetic maps are similar immediately surrounding the dfw locus, but show marked differences in other areas. A yeast artificial chromosome-based physical map suggests that the closest markers flanking the dfw locus, D6Mit11 (proximal) and Gat4, D6Mit55 (distal), are contained within 650-950 kb. The human homologues of the flanking loci Itpr1 (proximal) and Syn2 (distal) map to chromosome 3p25-p26, suggesting that the human homologue of the dfw gene is located within this same region.


Assuntos
Mapeamento Cromossômico , Camundongos Mutantes Neurológicos/genética , Animais , Ataxia/genética , Cromossomos Artificiais de Levedura , Cruzamentos Genéticos , Primers do DNA , Surdez/genética , Feminino , Marcadores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C3H , Repetições de Microssatélites , Reação em Cadeia da Polimerase , Recombinação Genética , Caracteres Sexuais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA